Adaptive lymphocyte profiles correlate to brain Aβ burden in patients with mild cognitive impairment

Ann M Stowe, Sara J Ireland, Sterling B Ortega, Ding Chen, Ryan M Huebinger, Takashi Tarumi, Thomas S Harris, C Munro Cullum, Roger Rosenberg, Nancy L Monson, Rong Zhang, Ann M Stowe, Sara J Ireland, Sterling B Ortega, Ding Chen, Ryan M Huebinger, Takashi Tarumi, Thomas S Harris, C Munro Cullum, Roger Rosenberg, Nancy L Monson, Rong Zhang

Abstract

Background: We previously found that subjects with amnestic mild cognitive impairment exhibit a pro-inflammatory immune profile in the cerebrospinal fluid similar to multiple sclerosis, a central nervous system autoimmune disease. We therefore hypothesized that early neuroinflammation would reflect increases in brain amyloid burden during amnestic mild cognitive impairment.

Methods: Cerebrospinal fluid and blood samples were collected from 24 participants with amnestic mild cognitive impairment (12 men, 12 women; 66 ± 6 years; 0.5 Clinical Dementia Rating) enrolled in the AETMCI study. Analyses of cerebrospinal fluid and blood included immune profiling by multi-parameter flow cytometry, genotyping for apolipoprotein (APO)ε, and quantification of cytokine and immunoglobin levels. Amyloid (A)β deposition was determined by 18F-florbetapir positron emission tomography. Spearman rank order correlations were performed to assess simple linear correlation for parameters including amyloid imaging, central and peripheral immune cell populations, and protein cytokine levels.

Results: Soluble Aβ42 in the cerebrospinal fluid declined as Aβ deposition increased overall and in the precuneous and posterior cingulate cortices. Lymphocyte profiling revealed a significant decline in T cell populations in the cerebrospinal fluid, specifically CD4+ T cells, as Aβ deposition in the posterior cingulate cortex increased. In contrast, increased Aβ burden correlated positively with increased memory B cells in the cerebrospinal fluid, which was exacerbated in APOε4 carriers. For peripheral circulating lymphocytes, only B cell populations decreased with Aβ deposition in the precuneous cortex, as peripheral T cell populations did not correlate with changes in brain amyloid burden.

Conclusions: Elevations in brain Aβ burden associate with a shift from T cells to memory B cells in the cerebrospinal fluid of subjects with amnestic mild cognitive impairment in this exploratory cohort. These data suggest the presence of cellular adaptive immune responses during Aβ accumulation, but further study needs to determine whether lymphocyte populations contribute to, or result from, Aβ dysregulation during memory decline on a larger cohort collected at multiple centers.

Trial registration: AETMCI NCT01146717.

Keywords: 18F-florbetapir; Amnestic mild cognitive impairment; Amyloid burden; CD4 T cells; Cerebrospinal fluid; IgG; Memory B cells; T lymphocytes.

Conflict of interest statement

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
PET reflects dysregulation of Aβ clearance. a, b Representative PET images and values corresponding to the standardized uptake value ratio (SUVR), with image of the posterior cingulate cortex (PCC). c Increases in PCC SUVR predict total amyloid burden (mean cortex). d PCC SUVR increase corresponds to a decrease in soluble CSF Aβ42. e, f Increases in precuneous cortex SUVR also predict total amyloid burden, but without the significant decrease in soluble CSF Aβ42. Linear regression (solid lines) and 95% confidence interval (dotted lines) shown. *p < 0.05, ***p < 0.001
Fig. 2
Fig. 2
T cell numbers in the CSF decrease with increased amyloid burden. Both the a total number of T cells in the cerebrospinal fluid (CSF) and b the number of CD4+ T cells in the CSF decreases as amyloid burden identified by PET standardized uptake value ratio (SUVR) in the posterior cingulate cortex (PCC) increases. c, d APOε status does not affect T cell populations numbers or percent representation in the CSF. Linear regression (solid lines) and 95% confidence interval (dotted lines) shown. *p < 0.05
Fig. 3
Fig. 3
B cells shift from the periphery to the CSF as amyloid burden increases. a Soluble IgG in the cerebrospinal fluid (CSF) declines with loss of soluble CSF Aβ42. b Increased mean standardized uptake value ratio (SUVR) for the total cortex correlates to an increase in memory B cell percent representation in the CSF, while c increased Aβ deposition in the precuneous cortex associates with a decline in B cell representation in the peripheral blood. d Soluble Aβ42 in the cerebrospinal fluid (CSF) is decreased in APOε4+ subjects. e, f Apoε4 carriers exhibit enhanced memory B cell percent representation in the CSF, but genotype did not affect overall B cell distribution
Fig. 4
Fig. 4
Summary of adaptive immune changes in the CSF with amyloid deposition. This figure highlights the findings that CSF lymphocyte populations shift from high CD4 T cells to high memory B cells with increased Aβ deposition in the brain. This also includes a reversal the IgG:soluble Aβ42 levels with increased amyloid burden

References

    1. Middleton LE, et al. Promising strategies for the prevention of dementia. Arch Neurol. 2009;66(10):1210–1215. doi: 10.1001/archneurol.2009.201.
    1. Richard E, et al. Methodological challenges in designing dementia prevention trials—the European Dementia Prevention Initiative (EDPI) J Neurol Sci. 2012;322(1-2):64–70. doi: 10.1016/j.jns.2012.06.012.
    1. Kling MA, et al. Vascular disease and dementias: paradigm shifts to drive research in new directions. Alzheimers Dement. 2013;9(1):76–92. doi: 10.1016/j.jalz.2012.02.007.
    1. Morris JK, et al. Is Alzheimer’s disease a systemic disease?, in Biochim Biophys Acta. 2014;1842(9):1340–1349.
    1. Rodrigue KM, et al. beta-Amyloid burden in healthy aging: regional distribution and cognitive consequences. Neurology. 2012;78(6):387–395. doi: 10.1212/WNL.0b013e318245d295.
    1. Villemagne VL, et al. Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer’s disease: a prospective cohort study. Lancet Neurol. 2013;12(4):357–367. doi: 10.1016/S1474-4422(13)70044-9.
    1. Storandt M, et al. Cognitive decline and brain volume loss as signatures of cerebral amyloid-beta peptide deposition identified with Pittsburgh compound B: cognitive decline associated with Abeta deposition. Arch Neurol. 2009;66(12):1476–1481. doi: 10.1001/archneurol.2009.272.
    1. Mawuenyega KG, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science. 2010;330(6012):1774. doi: 10.1126/science.1197623.
    1. Iwata N, et al. Identification of the major Abeta1-42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nat Med. 2000;6(2):143–150. doi: 10.1038/72237.
    1. Shibata M, et al. Clearance of Alzheimer’s amyloid-ss (1–40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest. 2000;106(12):1489–1499. doi: 10.1172/JCI10498.
    1. Iliff JJ, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4(147):147ra111.
    1. Akiyama H, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging. 2000;21(3):383–421. doi: 10.1016/S0197-4580(00)00124-X.
    1. Wyss-Coray T, et al. Inflammation in neurodegenerative disease—a double-edged sword. Neuron. 2002;35(3):419–432. doi: 10.1016/S0896-6273(02)00794-8.
    1. Querfurth HW, et al. Alzheimer’s disease. N Engl J Med. 2010;362(4):329–344. doi: 10.1056/NEJMra0909142.
    1. Tierney MC, et al. Prediction of probable Alzheimer’s disease in memory-impaired patients: a prospective longitudinal study. Neurology. 1996;46(3):661–665. doi: 10.1212/WNL.46.3.661.
    1. Bowen J, et al. Progression to dementia in patients with isolated memory loss. Lancet. 1997;349(9054):763–765. doi: 10.1016/S0140-6736(96)08256-6.
    1. Monson NL, et al. Elevated CNS inflammation in patients with preclinical Alzheimer’s disease. J Cereb Blood Flow Metab. 2014;34(1):30–33. doi: 10.1038/jcbfm.2013.183.
    1. Larbi A, et al. Dramatic shifts in circulating CD4 but not CD8 T cell subsets in mild Alzheimer’s disease. J Alzheimers Dis. 2009;17(1):91–103. doi: 10.3233/JAD-2009-1015.
    1. Pellicano M, et al. Immune profiling of Alzheimer patients. J Neuroimmunol. 2012;242(1–2):52–59. doi: 10.1016/j.jneuroim.2011.11.005.
    1. Speciale L, et al. Lymphocyte subset patterns and cytokine production in Alzheimer’s disease patients. Neurobiol Aging. 2007;28(8):1163–1169. doi: 10.1016/j.neurobiolaging.2006.05.020.
    1. Lombardi VR, et al. Characterization of cytokine production, screening of lymphocyte subset patterns and in vitro apoptosis in healthy and Alzheimer’s disease (AD) individuals. J Neuroimmunol. 1999;97(1–2):163–171. doi: 10.1016/S0165-5728(99)00046-6.
    1. Richartz-Salzburger E, et al. Altered lymphocyte distribution in Alzheimer’s disease. J Psychiatr Res. 2007;41(1–2):174–178. doi: 10.1016/j.jpsychires.2006.01.010.
    1. Clark CM, et al. Use of florbetapir-PET for imaging beta-amyloid pathology. JAMA. 2011;305(3):275–283. doi: 10.1001/jama.2010.2008.
    1. Selkoe DJ. Preventing Alzheimer’s disease. Science. 2012;337(6101):1488–1492. doi: 10.1126/science.1228541.
    1. Tarumi T, et al. Amyloid burden and sleep blood pressure in amnestic mild cognitive impairment. Neurology. 2015;85(22):1922–1929. doi: 10.1212/WNL.0000000000002167.
    1. Bero AW, et al. Neuronal activity regulates the regional vulnerability to amyloid-beta deposition. Nat Neurosci. 2011;14(6):750–756. doi: 10.1038/nn.2801.
    1. Buckner RL, et al. Molecular, structural, and functional characterization of Alzheimer’s disease: evidence for a relationship between default activity, amyloid, and memory. J Neurosci. 2005;25(34):7709–7717. doi: 10.1523/JNEUROSCI.2177-05.2005.
    1. Hatashita S, et al. Diagnosed mild cognitive impairment due to Alzheimer’s disease with PET biomarkers of beta amyloid and neuronal dysfunction. Plos One. 2013;8(6):e66877. doi: 10.1371/journal.pone.0066877.
    1. Lueg G, et al. Clinical relevance of specific T-cell activation in the blood and cerebrospinal fluid of patients with mild Alzheimer’s disease. Neurobiol Aging. 2015;36(1):81–89. doi: 10.1016/j.neurobiolaging.2014.08.008.
    1. Adluru N, et al. White matter microstructure in late middle-age: effects of apolipoprotein E4 and parental family history of Alzheimer’s disease. Neuroimage Clin. 2014;4:730–742. doi: 10.1016/j.nicl.2014.04.008.
    1. Holtzman DM, et al. Apolipoprotein E and apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harb Perspect Med. 2012;2(3):a006312. doi: 10.1101/cshperspect.a006312.
    1. Taylor JL, et al. APOE-epsilon4 and aging of medial temporal lobe gray matter in healthy adults older than 50 years. Neurobiol Aging. 2014;35(11):2479–2485. doi: 10.1016/j.neurobiolaging.2014.05.011.
    1. Fryer JD, et al. The low density lipoprotein receptor regulates the level of central nervous system human and murine apolipoprotein E but does not modify amyloid plaque pathology in PDAPP mice. J Biol Chem. 2005;280(27):25754–25759. doi: 10.1074/jbc.M502143200.
    1. De Sanctis JB, et al. Expression of low-density lipoprotein receptors in peripheral blood and tonsil B lymphocytes. Clin Exp Immunol. 1998;113(2):206–212. doi: 10.1046/j.1365-2249.1998.00579.x.
    1. Cirrito JR, et al. Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo. Neuron. 2005;48(6):913–922. doi: 10.1016/j.neuron.2005.10.028.
    1. Pfefferbaum A, et al. Cerebral blood flow in posterior cortical nodes of the default mode network decreases with task engagement but remains higher than in most brain regions. Cereb Cortex. 2011;21(1):233–244. doi: 10.1093/cercor/bhq090.
    1. Jozwik A, et al. Beta-amyloid peptides enhance the proliferative response of activated CD4CD28 lymphocytes from Alzheimer disease patients and from healthy elderly. Plos One. 2012;7(3):e33276. doi: 10.1371/journal.pone.0033276.
    1. Shlomchik MJ, et al. Germinal center selection and the development of memory B and plasma cells. Immunol Rev. 2012;247(1):52–63. doi: 10.1111/j.1600-065X.2012.01124.x.
    1. Ligocki AJ, et al. Expansion of CD27high plasmablasts in transverse myelitis patients that utilize VH4 and JH6 genes and undergo extensive somatic hypermutation. Genes Immun. 2013;14(5):291–301. doi: 10.1038/gene.2013.18.
    1. Metcalf TU, et al. Alphavirus-induced encephalomyelitis: antibody-secreting cells and viral clearance from the nervous system. J Virol. 2011;85(21):11490–11501. doi: 10.1128/JVI.05379-11.
    1. Tarawneh R, et al. Critical issues for successful immunotherapy in Alzheimer’s disease: development of biomarkers and methods for early detection and intervention. CNS Neurol Disord Drug Targets. 2009;8(2):144–159. doi: 10.2174/187152709787847324.
    1. Zotova E, et al. Inflammatory components in human Alzheimer’s disease and after active amyloid-beta42 immunization. Brain. 2013;136(Pt 9):2677–2696. doi: 10.1093/brain/awt210.
    1. Wisniewski T, et al. Immunotherapy for Alzheimer’s disease. Biochem Pharmacol. 2014;88(4):499–507. doi: 10.1016/j.bcp.2013.12.020.
    1. Bancos S, et al. Memory B cells from older people express normal levels of cyclooxygenase-2 and produce higher levels of IL-6 and IL-10 upon in vitro activation. Cell Immunol. 2010;266(1):90–97. doi: 10.1016/j.cellimm.2010.09.002.
    1. Drzezga A, et al. Effect of APOE genotype on amyloid plaque load and gray matter volume in Alzheimer disease. Neurology. 2009;72(17):1487–1494. doi: 10.1212/WNL.0b013e3181a2e8d0.
    1. Zhang H, et al. Cross-talk between apolipoprotein E and cytokines. Mediators Inflamm. 2011;2011:949072.
    1. Gale SC, et al. APOepsilon4 is associated with enhanced in vivo innate immune responses in human subjects. J Allergy Clin Immunol. 2014;134(1):127–34.
    1. Grocott HP, et al. Apolipoprotein E genotype differentially influences the proinflammatory and anti-inflammatory response to cardiopulmonary bypass. J Thorac Cardiovasc Surg. 2001;122(3):622–623. doi: 10.1067/mtc.2001.115152.
    1. Le Page A, et al. NK cells are activated in amnestic mild cognitive impairment but not in mild Alzheimer’s disease patients. J Alzheimers Dis. 2015;46(1):93–107. doi: 10.3233/JAD-143054.
    1. Fiala M, et al. Phagocytosis of amyloid-beta and inflammation: two faces of innate immunity in Alzheimer’s disease. J Alzheimers Dis. 2007;11(4):457–463. doi: 10.3233/JAD-2007-11406.
    1. Ligocki AJ, et al. A distinct class of antibodies may be an indicator of gray matter autoimmunity in early and established relapsing remitting multiple sclerosis patients. ASN Neuro. 2015;7(5):1–16. doi: 10.1177/1759091415609613.
    1. Schwartz M, et al. Breaking peripheral immune tolerance to CNS antigens in neurodegenerative diseases: boosting autoimmunity to fight-off chronic neuroinflammation. J Autoimmun. 2014;54:8–14. doi: 10.1016/j.jaut.2014.08.002.
    1. Musiek ES, et al. Three dimensions of the amyloid hypothesis: time, space and ‘wingmen’. Nat Neurosci. 2015;18(6):800–806. doi: 10.1038/nn.4018.
    1. Herrup K. The case for rejecting the amyloid cascade hypothesis. Nat Neurosci. 2015;18(6):794–799. doi: 10.1038/nn.4017.

Source: PubMed

3
Tilaa